Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Sci Data ; 11(1): 44, 2024 Jan 06.
Article in English | MEDLINE | ID: mdl-38184639

ABSTRACT

Mammalian energy homeostasis is primarilly regulated by the hypothalamus and hindbrain, with the hippocampus, midbrain nuclei, and other regions implicated by evidence from human genetics studies. To understand how these non-canonical brain regions respond to imbalances in energy homeostasis, we performed two experiments examining the effects of different diets in male C57BL6 mice. In our first study, groups of six pair-housed mice were given access to chow, high-fat diet or fasted for 16 hours. In our subsequent study, two groups of 10 mice were single-housed and given access to chow or fasted for 24 h. We recorded food intake for each cage, the change in body weight for each animal, and collected hypothalamus, hippocampus, superior colliculus, inferior colliculus, frontal cortex, and zona incerta-centric samples. We performed bulk RNA sequencing on 185 samples and validated them by a series of quality control assessments including alignment quality and gene expression profiling. We believe these studies capture the transcriptomic effects of acute fasting and high-fat diet in the rodent brain and provide a valuable reference.


Subject(s)
Fasting , Obesity , RNA-Seq , Animals , Male , Mice , Brain , Diet , Mice, Inbred C57BL , Obesity/genetics
2.
Elife ; 122023 09 12.
Article in English | MEDLINE | ID: mdl-37698546

ABSTRACT

Infection with Influenza A virus (IAV) causes the well-known symptoms of the flu, including fever, loss of appetite, and excessive sleepiness. These responses, mediated by the brain, will normally disappear once the virus is cleared from the system, but a severe respiratory virus infection may cause long-lasting neurological disturbances. These include encephalitis lethargica and narcolepsy. The mechanisms behind such long lasting changes are unknown. The hypothalamus is a central regulator of the homeostatic response during a viral challenge. To gain insight into the neuronal and non-neuronal molecular changes during an IAV infection, we intranasally infected mice with an H1N1 virus and extracted the brain at different time points. Using single-nucleus RNA sequencing (snRNA-seq) of the hypothalamus, we identify transcriptional effects in all identified cell populations. The snRNA-seq data showed the most pronounced transcriptional response at 3 days past infection, with a strong downregulation of genes across all cell types. General immune processes were mainly impacted in microglia, the brain resident immune cells, where we found increased numbers of cells expressing pro-inflammatory gene networks. In addition, we found that most neuronal cell populations downregulated genes contributing to the energy homeostasis in mitochondria and protein translation in the cytosol, indicating potential reduced cellular and neuronal activity. This might be a preventive mechanism in neuronal cells to avoid intracellular viral replication and attack by phagocytosing cells. The change of microglia gene activity suggest that this is complemented by a shift in microglia activity to provide increased surveillance of their surroundings.


When you are ill, your behaviour changes. You sleep more, eat less and are less likely to go out and be active. This behavioural change is called the 'sickness response' and is believed to help the immune system fight infection. An area of the brain called the hypothalamus helps to regulate sleep and appetite. Previous research has shown that when humans are ill, the immune system sends signals to the hypothalamus, likely initiating the sickness response. However, it was not clear which brain cells in the hypothalamus are involved in the response and how long after infection the brain returns to its normal state. To better understand the sickness response, Lemcke et al. infected mice with influenza then extracted and analysed brain tissue at different timepoints. The experiments showed that the major changes to gene expression in the hypothalamus early during an influenza infection are not happening in neurons ­ the cells in the brain that transmit electrical signals and usually control behaviour. Instead, it is cells called glia ­ which provide support and immune protection to the neurons ­ that change during infection. The findings suggest that these cells prepare to protect the neurons from influenza should the virus enter the brain. Lemcke et al. also found that the brain takes a long time to go back to normal after an influenza infection. In infected mice, molecular changes in brain cells could be detected even after the influenza infection had been cleared from the respiratory system. In the future, these findings may help to explain why some people take longer than others to fully recover from viral infections such as influenza and aid development of medications that speed up recovery.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza, Human , Animals , Mice , Humans , Hypothalamus , Solitary Nucleus , Appetite
3.
Cell Mol Life Sci ; 78(21-22): 6963-6978, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34586443

ABSTRACT

The endogenous chemokines CCL19 and CCL21 signal via their common receptor CCR7. CCL21 is the main lymph node homing chemokine, but a weak chemo-attractant compared to CCL19. Here we show that the 41-amino acid positively charged peptide, released through C-terminal cleavage of CCL21, C21TP, boosts the immune cell recruiting activity of CCL21 by up to 25-fold and the signaling activity via CCR7 by ~ 100-fold. Such boosting is unprecedented. Despite the presence of multiple basic glycosaminoglycan (GAG) binding motifs, C21TP boosting of CCL21 signaling does not involve interference with GAG mediated cell-surface retention. Instead, boosting is directly dependent on O-glycosylations in the CCR7 N-terminus. As dictated by the two-step binding model, the initial chemokine binding involves interaction of the chemokine fold with the receptor N-terminus, followed by insertion of the chemokine N-terminus deep into the receptor binding pocket. Our data suggest that apart from a role in initial chemokine binding, the receptor N-terminus also partakes in a gating mechanism, which could give rise to a reduced ligand activity, presumably through affecting the ligand positioning. Based on experiments that support a direct interaction of C21TP with the glycosylated CCR7 N-terminus, we propose that electrostatic interactions between the positively charged peptide and sialylated O-glycans in CCR7 N-terminus may create a more accessible version of the receptor and thus guide chemokine docking to generate a more favorable chemokine-receptor interaction, giving rise to the peptide boosting effect.


Subject(s)
Chemokine CCL21/metabolism , Dendritic Cells/metabolism , Lymph Nodes/metabolism , Receptors, CCR7/metabolism , Receptors, Lymphocyte Homing/metabolism , Signal Transduction/physiology , Animals , CHO Cells , Cells, Cultured , Cricetulus , Glycosylation , Humans , Ligands , Peptides/metabolism , Protein Binding/physiology , Static Electricity
4.
Diabetes ; 70(9): 1945-1955, 2021 09.
Article in English | MEDLINE | ID: mdl-34176785

ABSTRACT

The dorsal vagal complex (DVC) in the hindbrain, composed of the area postrema, nucleus of the solitary tract, and dorsal motor nucleus of the vagus, plays a critical role in modulating satiety. The incretins glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) act directly in the brain to modulate feeding, and receptors for both are expressed in the DVC. Given the impressive clinical responses to pharmacologic manipulation of incretin signaling, understanding the central mechanisms by which incretins alter metabolism and energy balance is of critical importance. Here, we review recent single-cell approaches used to detect molecular signatures of GLP-1 and GIP receptor-expressing cells in the DVC. In addition, we discuss how current advancements in single-cell transcriptomics, epigenetics, spatial transcriptomics, and circuit mapping techniques have the potential to further characterize incretin receptor circuits in the hindbrain.


Subject(s)
Glucagon-Like Peptide-1 Receptor/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Rhombencephalon/metabolism , Animals , Feeding Behavior/physiology , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Humans , Single-Cell Analysis
5.
Mol Metab ; 51: 101231, 2021 09.
Article in English | MEDLINE | ID: mdl-33831593

ABSTRACT

OBJECTIVE: GPR64/ADGRG2 is an orphan Adhesion G protein-coupled receptor (ADGR) known to be mainly expressed in the parathyroid gland and epididymis. This investigation aimed to delineate the cellular expression of GPR64 throughout the body with focus on the gastrointestinal (GI) tract. METHODS: Transgenic Gpr64mCherry reporter mice were histologically examined throughout the body and reporter protein expression in intestinal tuft cells was confirmed by specific cell ablation. The GPCR repertoire of intestinal Gpr64mCherry-positive tuft cells was analyzed by quantitative RT-PCR analysis and in situ hybridization. The Gpr64mCherry was crossed into the general tuft cell reporter Trpm5GFP to generate small intestinal organoids for time-lapse imaging. Intestinal tuft cells were isolated from small intestine, FACS-purified and transcriptionally compared using RNA-seq analysis. RESULTS: Expression of the Gpr64mCherry reporter was identified in multiple organs and specifically in olfactory microvillous cells, enteric nerves, and importantly in respiratory and GI tuft cells. In the small intestine, cell ablation targeting Gpr64-expressing epithelial cells eliminated tuft cells. Transcriptional analysis of small intestinal Gpr64mCherry -positive tuft cells confirmed expression of Gpr64 and the chemo-sensors Sucnr1, Gprc5c, Drd3, and Gpr41/Ffar3. Time-lapse studies of organoids from Trpm5GFP:Gpr64mCherry mice revealed sequential expression of initially Trpm5GFP and subsequently also Gpr64mCherry in maturing intestinal tuft cells. RNA-seq analysis of small intestinal tuft cells based on these two markers demonstrated a dynamic change in expression of transcription factors and GPCRs from young to mature tuft cells. CONCLUSIONS: GPR64 is expressed in chemosensory epithelial cells across a broad range of tissues; however, in the GI tract, GPR64 is remarkably selectively expressed in mature versus young immunoregulatory tuft cells.


Subject(s)
Chemoreceptor Cells/metabolism , Epithelial Cells/metabolism , Intestine, Small/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Female , Intestine, Small/cytology , Male , Mice , Mice, Transgenic , Receptors, G-Protein-Coupled/analysis , Receptors, G-Protein-Coupled/genetics
6.
Nat Metab ; 3(4): 530-545, 2021 04.
Article in English | MEDLINE | ID: mdl-33767443

ABSTRACT

The brainstem dorsal vagal complex (DVC) is known to regulate energy balance and is the target of appetite-suppressing hormones, such as glucagon-like peptide 1 (GLP-1). Here we provide a comprehensive genetic map of the DVC and identify neuronal populations that control feeding. Combining bulk and single-nucleus gene expression and chromatin profiling of DVC cells, we reveal 25 neuronal populations with unique transcriptional and chromatin accessibility landscapes and peptide receptor expression profiles. GLP-1 receptor (GLP-1R) agonist administration induces gene expression alterations specific to two distinct sets of Glp1r neurons-one population in the area postrema and one in the nucleus of the solitary tract that also expresses calcitonin receptor (Calcr). Transcripts and regions of accessible chromatin near obesity-associated genetic variants are enriched in the area postrema and the nucleus of the solitary tract neurons that express Glp1r and/or Calcr, and activating several of these neuronal populations decreases feeding in rodents. Thus, DVC neuronal populations associated with obesity predisposition suppress feeding and may represent therapeutic targets for obesity.


Subject(s)
Chromosome Mapping , Obesity/genetics , Obesity/physiopathology , Vagus Nerve/physiopathology , Animals , Appetite/genetics , Body Weight/genetics , Brain Stem/physiopathology , Calcitonin Receptor-Like Protein/genetics , Cell Nucleus/genetics , Chromatin/genetics , Chromatin/metabolism , Gene Expression , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons , Solitary Nucleus/physiology
7.
Am J Physiol Endocrinol Metab ; 320(5): E874-E885, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33645250

ABSTRACT

The aim of this study was to explore individual amino acid-stimulated GLP-1 responses and the underlying stimulatory mechanisms, as well as to identify the amino acid-sensing receptors involved in amino acid-stimulated GLP-1 release. Experiments were primarily based on isolated perfused rat small intestines, which have intact epithelial polarization allowing discrimination between luminal and basolateral mechanisms as well as quantitative studies of intestinal absorption and hormone secretion. Expression analysis of amino acid sensors on isolated murine GLP-1 secreting L-cells was assessed by qPCR. We found that l-valine powerfully stimulated GLP-1 secretion but only from the luminal side (2.9-fold increase). When administered from the vascular side, l-arginine and the aromatic amino acids stimulated GLP-1 secretion equally (2.6- to 2.9-fold increases). Expression analysis revealed that Casr expression was enriched in murine GLP-1 secreting L-cells, whereas Gpr35, Gprc6a, Gpr142, Gpr93 (Lpar5), and the umami taste receptor subunits Tas1r3 and Tas1r1 were not. Consistently, activation of GPR35, GPR93, GPR142, and the umami taste receptor with specific agonists or allosteric modulators did not increase GLP-1 secretion (P > 0.05 for all experiments), whereas vascular inhibition of CaSR reduced GLP-1 secretion in response to luminal infusion of mixed amino acids. In conclusion, amino acids differ in their capacity to stimulate GLP-1 secretion. Some amino acids stimulated secretion only from the intestinal lumen, whereas other amino acids exclusively stimulated secretion from the vascular side, indicating that amino acid-stimulated GLP-1 secretion involves both apical and basolateral (postabsorptive) sensing mechanisms. Sensing of absorbed amino acids involves CaSR activation as vascular inhibition of CaSR markedly diminished amino acid stimulated GLP-1 release.NEW & NOTEWORTHY Using isolated perfused rat small intestines, we show that amino acids differ in their mechanisms and capacity of stimulating GLP-1 release. Furthermore, we demonstrate that sensing by GPR142, GPR35, GPR93, and the umami taste receptor (Tas1R1/Tas1R3) are not involved in amino acid stimulated GLP-1 release. In contrast to previous studies, this experimental model allows discrimination between the luminal and the vascular side of the intestine, which is essential when studying mechanisms of amino acid-stimulated GLP-1 secretion.


Subject(s)
Amino Acids/pharmacology , Glucagon-Like Peptide 1/metabolism , Intestine, Small/drug effects , Animals , Intestine, Small/metabolism , Intestine, Small/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Perfusion , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Receptors, Lysophosphatidic Acid/agonists , Receptors, Lysophosphatidic Acid/metabolism , Secretory Pathway/drug effects , Signal Transduction/drug effects
8.
Nat Commun ; 11(1): 4458, 2020 09 07.
Article in English | MEDLINE | ID: mdl-32895383

ABSTRACT

In rodent models of type 2 diabetes (T2D), sustained remission of hyperglycemia can be induced by a single intracerebroventricular (icv) injection of fibroblast growth factor 1 (FGF1), and the mediobasal hypothalamus (MBH) was recently implicated as the brain area responsible for this effect. To better understand the cellular response to FGF1 in the MBH, we sequenced >79,000 single-cell transcriptomes from the hypothalamus of diabetic Lepob/ob mice obtained on Days 1 and 5 after icv injection of either FGF1 or vehicle. A wide range of transcriptional responses to FGF1 was observed across diverse hypothalamic cell types, with glial cell types responding much more robustly than neurons at both time points. Tanycytes and ependymal cells were the most FGF1-responsive cell type at Day 1, but astrocytes and oligodendrocyte lineage cells subsequently became more responsive. Based on histochemical and ultrastructural evidence of enhanced cell-cell interactions between astrocytes and Agrp neurons (key components of the melanocortin system), we performed a series of studies showing that intact melanocortin signaling is required for the sustained antidiabetic action of FGF1. These data collectively suggest that hypothalamic glial cells are leading targets for the effects of FGF1 and that sustained diabetes remission is dependent on intact melanocortin signaling.


Subject(s)
Diabetes Mellitus, Experimental/diet therapy , Diabetes Mellitus, Type 2/drug therapy , Fibroblast Growth Factor 1/administration & dosage , Hypoglycemic Agents/administration & dosage , Hypothalamus/drug effects , Recombinant Proteins/administration & dosage , Agouti-Related Protein/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Blood Glucose/analysis , Cell Communication , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat/adverse effects , Dietary Sucrose/administration & dosage , Dietary Sucrose/adverse effects , Humans , Hypothalamus/cytology , Hypothalamus/pathology , Injections, Intraventricular , Leptin/genetics , Male , Melanocortins/metabolism , Melanocyte-Stimulating Hormones/administration & dosage , Mice , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , RNA-Seq , Receptor, Melanocortin, Type 4/genetics , Receptors, Melanocortin/antagonists & inhibitors , Receptors, Melanocortin/metabolism , Remission Induction/methods , Signal Transduction/drug effects , Single-Cell Analysis , Stereotaxic Techniques , Transcriptome/drug effects
10.
Nat Biotechnol ; 38(11): 1265-1273, 2020 11.
Article in English | MEDLINE | ID: mdl-32451506

ABSTRACT

The study of brain development in humans is limited by the lack of tissue samples and suitable in vitro models. Here, we model early human neural tube development using human embryonic stem cells cultured in a microfluidic device. The approach, named microfluidic-controlled stem cell regionalization (MiSTR), exposes pluripotent stem cells to signaling gradients that mimic developmental patterning. Using a WNT-activating gradient, we generated a neural tissue exhibiting progressive caudalization from forebrain to midbrain to hindbrain, including formation of isthmic organizer characteristics. Single-cell transcriptomics revealed that rostro-caudal organization was already established at 24 h of differentiation, and that the first markers of a neural-specific transcription program emerged in the rostral cells at 48 h. The transcriptomic hallmarks of rostro-caudal organization recapitulated gene expression patterns of the early rostro-caudal neural plate in mouse embryos. Thus, MiSTR will facilitate research on the factors and processes underlying rostro-caudal neural tube patterning.


Subject(s)
Cell Differentiation , Human Embryonic Stem Cells/cytology , Microfluidics/methods , Neural Tube/embryology , Wnt Proteins/metabolism , Body Patterning , Cells, Cultured , Gene Expression Regulation, Developmental , Humans , Single-Cell Analysis , Transcriptome/genetics , Wnt Signaling Pathway
11.
Diabetes ; 69(4): 614-623, 2020 04.
Article in English | MEDLINE | ID: mdl-32041793

ABSTRACT

Glucagon-like peptide 1 (GLP-1) mimetics are effective drugs for treatment of type 2 diabetes, and there is consequently extensive interest in increasing endogenous GLP-1 secretion and L-cell abundance. Here we identify G-protein-coupled bile acid receptor 1 (GPBAR1) as a selective regulator of intestinal L-cell differentiation. Lithocholic acid and the synthetic GPBAR1 agonist, L3740, selectively increased L-cell density in mouse and human intestinal organoids and elevated GLP-1 secretory capacity. L3740 induced expression of Gcg and transcription factors Ngn3 and NeuroD1 L3740 also increased the L-cell number and GLP-1 levels and improved glucose tolerance in vivo. Further mechanistic examination revealed that the effect of L3740 on L cells required intact GLP-1 receptor and serotonin 5-hydroxytryptamine receptor 4 (5-HT4) signaling. Importantly, serotonin signaling through 5-HT4 mimicked the effects of L3740, acting downstream of GLP-1. Thus, GPBAR1 agonists and other powerful GLP-1 secretagogues facilitate L-cell differentiation through a paracrine GLP-1-dependent and serotonin-mediated mechanism.


Subject(s)
Bile Acids and Salts/pharmacology , Cell Differentiation/drug effects , Enteroendocrine Cells/drug effects , Glucagon-Like Peptide 1/metabolism , Paracrine Communication/physiology , Receptors, G-Protein-Coupled/metabolism , Serotonin/metabolism , Animals , Enteroendocrine Cells/physiology , Female , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Jejunum/drug effects , Jejunum/metabolism , Male , Mice , Paracrine Communication/drug effects , Signal Transduction/drug effects
12.
Trends Neurosci ; 42(10): 663-666, 2019 10.
Article in English | MEDLINE | ID: mdl-31421944

ABSTRACT

Using single-cell RNA sequencing (RNA-seq), Kupari and coworkers (Cell Rep., 2019) have generated a long sought-after molecular atlas of vagal afferents in the mouse. Vagal afferents were found to be organized into 24 subtypes, revealing a level of diversity that was not previously recognized.


Subject(s)
Sensory Receptor Cells , Vagus Nerve , Animals , Mice
13.
J Neuroendocrinol ; 31(7): e12761, 2019 07.
Article in English | MEDLINE | ID: mdl-31237372

ABSTRACT

Dopamine-producing tyrosine hydroxylase (TH) neurones in the hypothalamic arcuate nucleus (ARC) have recently been shown to be involved in ghrelin signalling and body weight homeostasis. In the present study, we investigate the role of the intracellular regulator RhoA in hypothalamic TH neurones in response to peripheral hormones. Diet-induced obesity was found to be associated with increased phosphorylation of TH in ARC, indicating obesity-associated increased activity of ARC TH neurones. Mice in which RhoA was specifically knocked out in TH neurones (TH-RhoA-/- mice) were more sensitive to the orexigenic effect of peripherally administered ghrelin and displayed an abolished response to the anorexigenic hormone leptin. When TH-RhoA-/- mice were challenged with a high-fat high-sucrose (HFHS) diet, they became hyperphagic and gained more body weight and fat mass compared to wild-type control mice. Importantly, lack of RhoA prevented development of ghrelin resistance, which is normally observed in wild-type mice after long-term HFHS diet feeding. Patch-clamp electrophysiological analysis demonstrated increased ghrelin-induced excitability of TH neurones in lean TH-RhoA-/- mice compared to lean littermate control animals. Additionally, increased expression of the orexigenic hypothalamic neuropeptides agouti-related peptide and neuropeptide Y was observed in TH-RhoA-/- mice. Overall, our data indicate that TH neurones in ARC are important for the regulation of body weight homeostasis and that RhoA is both a central effector in these neurones and important for the development of obesity-induced ghrelin resistance. The obese phenotype of TH-RhoA-/- mice may be a result of increased sensitivity to ghrelin and decreased sensitivity to leptin, resulting in increased food intake.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Body Weight , Eating , Ghrelin/metabolism , Neurons/metabolism , Tyrosine 3-Monooxygenase/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Female , Gene Expression , Male , Mice, Knockout , Obesity/metabolism , RNA, Messenger/metabolism , rhoA GTP-Binding Protein/genetics
14.
Nat Commun ; 10(1): 2889, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31253831

ABSTRACT

The sinus node is a collection of highly specialised cells constituting the heart's pacemaker. The molecular underpinnings of its pacemaking abilities are debated. Using high-resolution mass spectrometry, we here quantify >7,000 proteins from sinus node and neighbouring atrial muscle. Abundances of 575 proteins differ between the two tissues. By performing single-nucleus RNA sequencing of sinus node biopsies, we attribute measured protein abundances to specific cell types. The data reveal significant differences in ion channels responsible for the membrane clock, but not in Ca2+ clock proteins, suggesting that the membrane clock underpins pacemaking. Consistently, incorporation of ion channel expression differences into a biophysically-detailed atrial action potential model result in pacemaking and a sinus node-like action potential. Combining our quantitative proteomics data with computational modeling, we estimate ion channel copy numbers for sinus node myocytes. Our findings provide detailed insights into the unique molecular make-up of the cardiac pacemaker.


Subject(s)
Biological Clocks/physiology , Peptides/chemistry , Peptides/metabolism , Proteomics , Sinoatrial Node/metabolism , Transcriptome , Action Potentials , Animals , Chromatography, Liquid , Gene Expression Regulation/physiology , Hydrogen-Ion Concentration , Male , Mice , Mice, Inbred C57BL , Peptides/genetics , Tandem Mass Spectrometry
15.
Mol Metab ; 19: 49-64, 2019 01.
Article in English | MEDLINE | ID: mdl-30472415

ABSTRACT

OBJECTIVES: GPR142, which is highly expressed in pancreatic islets, has recently been deorphanized as a receptor for aromatic amino acids; however, its physiological role and pharmacological potential is unclear. METHODS AND RESULTS: We find that GPR142 is expressed not only in ß- but also in α-cells of the islets as well as in enteroendocrine cells, and we confirm that GPR142 is a highly selective sensor of essential aromatic amino acids, in particular Trp and oligopeptides with N-terminal Trp. GPR142 knock-out mice displayed a very limited metabolic phenotype but demonstrated that L-Trp induced secretion of pancreatic and gut hormones is mediated through GPR142 but that the receptor is not required for protein-induced hormone secretion. A synthetic GPR142 agonist stimulated insulin and glucagon as well as GIP, CCK, and GLP-1 secretion. In particular, GIP secretion was sensitive to oral administration of the GPR142 agonist an effect which in contrast to the other hormones was blocked by protein load. Oral administration of the GPR142 agonist increased [3H]-2-deoxyglucose uptake in muscle and fat depots mediated through insulin action while it lowered liver glycogen conceivably mediated through glucagon, and, consequently, it did not lower total blood glucose. Nevertheless, acute administration of the GPR142 agonist strongly improved oral glucose tolerance in both lean and obese mice as well as Zucker fatty rat. Six weeks in-feed chronic treatment with the GPR142 agonist did not affect body weight in DIO mice, but increased energy expenditure and carbohydrate utilization, lowered basal glucose, and improved insulin sensitivity. CONCLUSIONS: GPR142 functions as a sensor of aromatic amino acids, controlling GIP but also CCK and GLP-1 as well as insulin and glucagon in the pancreas. GPR142 agonists could have novel interesting potential in modifying metabolism through a balanced action of gut hormones as well as both insulin and glucagon.


Subject(s)
Islets of Langerhans/metabolism , Receptors, G-Protein-Coupled/metabolism , Amino Acids, Aromatic/metabolism , Animals , Blood Glucose/metabolism , Glucagon/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Secreting Cells/metabolism , Glucose/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Rats , Rats, Zucker , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Glucagon/metabolism , Tryptophan/metabolism
16.
Am J Physiol Endocrinol Metab ; 316(3): E453-E463, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30562060

ABSTRACT

Dietary fibers, an integral part of the human diet, require the enzymatic activity of the gut microbiota for complete metabolism into short-chain fatty acids (SCFAs). SCFAs are important modulators of host metabolism and physiology and act in part as signaling molecules by activating G protein-coupled receptors (GPCRs), such as GPR41. Flaxseed fibers improve metabolism in rodents and mice, but their fermentation profiles, effects on enteroendocrine cells, and associated metabolic benefits are unknown. We fed GPR41-red fluorescent protein mice, an enteroendocrine reporter mouse strain, chow, high-fat diet (HFD), or HFD supplemented either with 10% nonfermentable fiber cellulose or fermentable flaxseed fibers for 12 wk to assess changes in cecal gut microbiota, enteroendocrine cell transcriptome in the ileum and colon, and physiological parameters. We observed that flaxseed fibers restructured the gut microbiota and promoted proliferation of the genera Bifidobacterium and Akkermansia compared with HFD. The shifts in cecal bacterial composition restored levels of the SCFAs butyrate similar to the chow diet, resulting in colonic but not ileal enteroendocrine cell transcriptional changes in genes related to cell cycle, mRNA, and protein transport compared with HFD. Consistent with the effects on enteroendocrine functions, flaxseed fibers also protected mice from diet-induced obesity, potentially by preventing a reduction in energy expenditure induced by an HFD. Our study shows that flaxseed fibers alter cecal microbial ecology, are fermented to SCFAs in the cecum, and modulate enteroendocrine cell transcriptome in the colon, which may contribute to their metabolically favorable phenotype.


Subject(s)
Enteroendocrine Cells/metabolism , Fatty Acids, Volatile/metabolism , Fermentation , Flax/metabolism , Gastrointestinal Microbiome , Obesity/metabolism , Transcriptome , Animals , Bifidobacterium , Cecum/microbiology , Cellulose , Colon/cytology , Diet, High-Fat , Dietary Fiber , Female , Firmicutes , Ileum/cytology , Lactobacillus , Male , Mice , Receptors, G-Protein-Coupled/metabolism , Verrucomicrobia
17.
Gastroenterology ; 155(4): 1164-1176.e2, 2018 10.
Article in English | MEDLINE | ID: mdl-29935151

ABSTRACT

BACKGROUND & AIMS: Glucagon-like peptide 1 (GLP1) is produced by L cells in the intestine, and agonists of the GLP1 receptor are effective in the treatment of diabetes. Levels of GLP1 increase with numbers of L cells. Therefore, agents that increase numbers of L cell might be developed for treatment of diabetes. Ras homologue family member A (RhoA) signaling through Rho-associated coiled-coil-containing protein kinases 1 and 2 (ROCK1 and ROCK2) controls cell differentiation, but it is not clear whether this pathway regulates enteroendocrine differentiation in the intestinal epithelium. We investigated the effects of Y-27632, an inhibitor of ROCK1 and ROCK2, on L-cell differentiation. METHODS: We collected intestinal tissues from GLU-Venus, GPR41-RFP, and Neurog3-RFP mice, in which the endocrine lineage is fluorescently labeled, for in vitro culture and histologic analysis. Small intestine organoids derived from these mice were cultured with Y-27632 and we measured percentages of L cells, expression of intestinal cell-specific markers, and secretion of GLP1 in medium. Mice were fed a normal chow or a high-fat diet and given Y-27632 or saline (control) and blood samples were collected for measurement of GLP1, insulin, and glucose. RESULTS: Incubation of intestinal organoids with Y-27632 increased numbers of L cells and secretion of GLP1. These increases were associated with upregulated expression of genes encoding intestinal hormones, neurogenin 3, neurogenic differentiation factor 1, forkhead box A1 and A2, and additional markers of secretory cells. Mice fed the normal chow diet and given Y-27632 had increased numbers of L cells in intestinal tissues, increased plasma levels of GLP1 and insulin, and lower blood levels of glucose compared with mice fed the normal chow diet and given saline. In mice with insulin resistance induced by the high-fat diet, administration of Y-27632 increased secretion of GLP1 and glucose tolerance compared with administration of saline. CONCLUSIONS: In mouse intestinal organoids, an inhibitor of RhoA signaling increased the differentiation of the secretory lineage and the development of enteroendocrine cells. Inhibitors of RhoA signaling or other strategies to increase numbers of L cells might be developed for treatment of patients with type 2 diabetes or for increasing glucose tolerance.


Subject(s)
Amides/pharmacology , Blood Glucose/drug effects , Cell Differentiation/drug effects , Enteroendocrine Cells/drug effects , Glucose Intolerance/drug therapy , Hypoglycemic Agents/pharmacology , Ileum/drug effects , Insulin Resistance , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Stem Cells/drug effects , rho GTP-Binding Proteins/metabolism , Animals , Biomarkers/blood , Blood Glucose/metabolism , Cell Lineage , Cell Proliferation/drug effects , Diet, High-Fat , Disease Models, Animal , Enteroendocrine Cells/metabolism , Glucagon-Like Peptide 1/blood , Glucose Intolerance/blood , Glucose Intolerance/etiology , Glucose Intolerance/physiopathology , Ileum/metabolism , Insulin/blood , Male , Mice, Inbred C57BL , Mice, Transgenic , Organoids/drug effects , Organoids/metabolism , Phenotype , Signal Transduction/drug effects , Stem Cells/metabolism , Time Factors , Tissue Culture Techniques , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein
18.
Mol Metab ; 12: 62-75, 2018 06.
Article in English | MEDLINE | ID: mdl-29673577

ABSTRACT

OBJECTIVES: G protein-coupled receptors (GPCRs) act as transmembrane molecular sensors of neurotransmitters, hormones, nutrients, and metabolites. Because unmyelinated vagal afferents richly innervate the gastrointestinal mucosa, gut-derived molecules may directly modulate the activity of vagal afferents through GPCRs. However, the types of GPCRs expressed in vagal afferents are largely unknown. Here, we determined the expression profile of all GPCRs expressed in vagal afferents of the mouse, with a special emphasis on those innervating the gastrointestinal tract. METHODS: Using a combination of high-throughput quantitative PCR, RNA sequencing, and in situ hybridization, we systematically quantified GPCRs expressed in vagal unmyelinated Nav1.8-expressing afferents. RESULTS: GPCRs for gut hormones that were the most enriched in Nav1.8-expressing vagal unmyelinated afferents included NTSR1, NPY2R, CCK1R, and to a lesser extent, GLP1R, but not GHSR and GIPR. Interestingly, both GLP1R and NPY2R were coexpressed with CCK1R. In contrast, NTSR1 was coexpressed with GPR65, a marker preferentially enriched in intestinal mucosal afferents. Only few microbiome-derived metabolite sensors such as GPR35 and, to a lesser extent, GPR119 and CaSR were identified in the Nav1.8-expressing vagal afferents. GPCRs involved in lipid sensing and inflammation (e.g. CB1R, CYSLTR2, PTGER4), and neurotransmitters signaling (CHRM4, DRD2, CRHR2) were also highly enriched in Nav1.8-expressing neurons. Finally, we identified 21 orphan GPCRs with unknown functions in vagal afferents. CONCLUSION: Overall, this study provides a comprehensive description of GPCR-dependent sensing mechanisms in vagal afferents, including novel coexpression patterns, and conceivably coaction of key receptors for gut-derived molecules involved in gut-brain communication.


Subject(s)
Brain/metabolism , Gastrointestinal Hormones/metabolism , Intestinal Mucosa/metabolism , Neurons, Afferent/metabolism , Receptors, G-Protein-Coupled/metabolism , Vagus Nerve/metabolism , Animals , Cells, Cultured , Intestinal Mucosa/innervation , Male , Mice , Mice, Inbred C57BL , NAV1.8 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Neurons, Afferent/physiology , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Vagus Nerve/physiology
19.
Mol Metab ; 11: 70-83, 2018 05.
Article in English | MEDLINE | ID: mdl-29576437

ABSTRACT

OBJECTIVES: 5-HT storing enterochromaffin (EC) cells are believed to respond to nutrient and gut microbial components, and 5-HT receptor-expressing afferent vagal neurons have been described to be the major sensors of nutrients in the GI-tract. However, the molecular mechanism through which EC cells sense nutrients and gut microbiota is still unclear. METHODS AND RESULTS: TPH1, the 5-HT generating enzyme, and chromogranin A, an acidic protein responsible for secretory granule storage of 5-HT, were highly enriched in FACS-purified EC cells from both small intestine and colon using a 5-HT antibody-based method. Surprisingly, EC cells from the small intestine did not express GPCR sensors for lipid and protein metabolites, such as FFAR1, GPR119, GPBAR1 (TGR5), CaSR, and GPR142, in contrast to the neighboring GLP-1 storing enteroendocrine cell. However, the GLP-1 receptor was particularly highly expressed and enriched in EC cells as judged both by qPCR and by immunohistochemistry using a receptor antibody. GLP-1 receptor agonists robustly stimulated 5-HT secretion from intestinal preparations using both HPLC and a specific amperometric method. Colonic EC cells expressed many different types of known and potential GPCR sensors of microbial metabolites including three receptors for SCFAs, i.e. FFAR2, OLF78, and OLF558 and receptors for aromatic acids, GPR35; secondary bile acids GPBAR1; and acyl-amides and lactate, GPR132. CONCLUSION: Nutrient metabolites apparently do not stimulate EC cells of the small intestine directly but through a paracrine mechanism involving GLP-1 secreted from neighboring enteroendocrine cells. In contrast, colonic EC cells are able to sense a multitude of different metabolites generated by the gut microbiota as well as gut hormones, including GLP-1.


Subject(s)
Enterochromaffin Cells/metabolism , Gastrointestinal Microbiome , Glucagon-Like Peptide 1/metabolism , Signal Transduction , Animals , Cells, Cultured , Chromogranin A/metabolism , Fatty Acids/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Male , Mice , Mice, Inbred C57BL , Serotonin/metabolism
20.
J Med Chem ; 60(3): 886-898, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28045522

ABSTRACT

The G-protein-coupled receptor 39 (GPR39) is a G-protein-coupled receptor activated by Zn2+. We used a homology model-based approach to identify small-molecule pharmacological tool compounds for the receptor. The method focused on a putative binding site in GPR39 for synthetic ligands and knowledge of ligand binding to other receptors with similar binding pockets to select iterative series of minilibraries. These libraries were cherry-picked from all commercially available synthetic compounds. A total of only 520 compounds were tested in vitro, making this method broadly applicable for tool compound development. The compounds of the initial library were inactive when tested alone, but lead compounds were identified using Zn2+ as an allosteric enhancer. Highly selective, highly potent Zn2+-independent GPR39 agonists were found in subsequent minilibraries. These agonists identified GPR39 as a novel regulator of gastric somatostatin secretion.


Subject(s)
Receptors, G-Protein-Coupled/agonists , Zinc/metabolism , Allosteric Regulation , Drug Discovery , Gastric Mucosa/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Molecular Structure , Receptors, G-Protein-Coupled/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...